Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
JNCI Cancer Spectr ; 5(5)2021 10.
Article in English | MEDLINE | ID: mdl-34485814

ABSTRACT

Background: Circulating levels of cancer antigen (CA) 15-3, a tumor marker and regulator of cellular metabolism, were reduced by metformin in a nonrandomized neoadjuvant study. We examined the effects of metformin (vs placebo) on CA 15-3 in participants of MA.32, a phase III randomized trial in early-stage breast cancer. Methods: A total of 3649 patients with T1-3, N0-3, M0 breast cancer were randomly assigned; pretreatment and 6-month on-treatment fasting plasma were centrally assayed for CA 15-3. Genomic DNA was analyzed for the rs11212617 single nucleotide polymorphism. Absolute and relative change of CA 15-3 (metformin vs placebo) were compared using Wilcoxon rank and t tests. Regression models adjusted for baseline differences and assessed key interactions. All statistical tests were 2-sided. Results: Mean (SD) age was 52.4 (10.0) years. The majority of patients had T2/3, node-positive, hormone receptor-positive, HER2-negative breast cancer treated with (neo)adjuvant chemotherapy and hormone therapy. Mean (SD) baseline CA 15-3 was 17.7 (7.6) and 18.0 (8.1 U/mL). At 6 months, CA 15-3 was statistically significantly reduced in metformin vs placebo arms (absolute geometric mean reduction in CA 15-3 = 7.7% vs 2.0%, P < .001; relative metformin: placebo level of CA 15-3 [adjusted for age, baseline body mass index, and baseline CA 15-3] = 0.94, 95% confidence interval = 0.92 to 0.96). This reduction was independent of tumor characteristics, perioperative systemic therapy, baseline body mass index, insulin, and the single nucleotide polymorphism status (all Ps > .11). Conclusions: Our observation that metformin reduces CA 15-3 by approximately 6% was corroborated in a large placebo-controlled randomized trial. The clinical implications of this reduction in CA 15-3 will be explored in upcoming efficacy analyses of breast cancer outcomes in MA.32.


Subject(s)
Breast Neoplasms/blood , Metformin/therapeutic use , Mucin-1/blood , Body Mass Index , Breast Neoplasms/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Fasting/blood , Female , Humans , Middle Aged , Mucin-1/drug effects , Placebos/therapeutic use , Polymorphism, Single Nucleotide
2.
NPJ Breast Cancer ; 7(1): 74, 2021 Jun 08.
Article in English | MEDLINE | ID: mdl-34103538

ABSTRACT

Metformin may exert anticancer effects through indirect (mediated by metabolic changes) or direct mechanisms. The goal was to examine metformin impact on metabolic factors in non-diabetic subjects and determine whether this impact varies by baseline BMI, insulin, and rs11212617 SNP in CCTG MA.32, a double-blind placebo-controlled randomized adjuvant breast cancer (BC) trial. 3649 subjects with T1-3, N0-3, M0 BC were randomized; pretreatment and 6-month on-treatment fasting plasma was centrally assayed for insulin, leptin, highly sensitive C-reactive protein (hsCRP). Glucose was measured locally and homeostasis model assessment (HOMA) calculated. Genomic DNA was analyzed for the rs11212617 SNP. Absolute and relative change of metabolic factors (metformin versus placebo) were compared using Wilcoxon rank and t-tests. Regression models were adjusted for baseline differences and assessed interactions with baseline BMI, insulin, and the SNP. Mean age was 52 years. The majority had T2/3, node positive, hormone receptor positive, HER2 negative BC treated with (neo)adjuvant chemotherapy and hormone therapy. Median baseline body mass index (BMI) was 27.4 kg/m2 (metformin) and 27.3 kg/m2 (placebo). Median weight change was -1.4 kg (metformin) vs +0.5 kg (placebo). Significant improvements were seen in all metabolic factors, with 6 month standardized ratios (metformin/placebo) of 0.85 (insulin), 0.83 (HOMA), 0.80 (leptin), and 0.84 (hsCRP), with no qualitative interactions with baseline BMI or insulin. Changes did not differ by rs11212617 allele. Metformin (vs placebo) led to significant improvements in weight and metabolic factors; these changes did not differ by rs11212617 allele status.

3.
J Natl Cancer Inst ; 113(2): 192-198, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33527137

ABSTRACT

BACKGROUND: Metformin has been associated with lower breast cancer (BC) risk and improved outcomes in observational studies. Multiple biologic mechanisms have been proposed, including a recent report of altered sex hormones. We evaluated the effect of metformin on sex hormones in MA.32, a phase III trial of nondiabetic BC subjects who were randomly assigned to metformin or placebo. METHODS: We studied the subgroup of postmenopausal hormone receptor-negative BC subjects not receiving endocrine treatment who provided fasting blood at baseline and at 6 months after being randomly assigned. Sex hormone-binding globulin, bioavailable testosterone, and estradiol levels were assayed using electrochemiluminescence immunoassay. Change from baseline to 6 months between study arms was compared using Wilcoxon sum rank tests and regression models. RESULTS: 312 women were eligible (141 metformin vs 171 placebo); the majority of subjects in each arm had T1/2, N0, HER2-negative BC and had received (neo)adjuvant chemotherapy. Mean age was 58.1 (SD=6.9) vs 57.5 (SD=7.9) years, mean body mass index (BMI) was 27.3 (SD=5.5) vs 28.9 (SD=6.4) kg/m2 for metformin vs placebo, respectively. Median estradiol decreased between baseline and 6 months on metformin vs placebo (-5.7 vs 0 pmol/L; P < .001) in univariable analysis and after controlling for baseline BMI and BMI change (P < .001). There was no change in sex hormone-binding globulin or bioavailable testosterone. CONCLUSION: Metformin lowered estradiol levels, independent of BMI. This observation suggests a new metformin effect that has potential relevance to estrogen sensitive cancers.


Subject(s)
Breast Neoplasms/drug therapy , Gonadal Steroid Hormones/antagonists & inhibitors , Metformin/administration & dosage , Body Mass Index , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Estradiol/genetics , Female , Gonadal Steroid Hormones/genetics , Humans , Middle Aged , Receptor, ErbB-2/genetics , Testosterone/antagonists & inhibitors , Testosterone/genetics
4.
JNCI Cancer Spectr ; 4(1): pkz097, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32206745
5.
Article in English | MEDLINE | ID: mdl-31712221

ABSTRACT

For years, clinical and basic researchers have been aware of the presence of PTEN in the nucleus in cell culture, animal models, and both healthy and diseased human tissues. Despite the early recognition of nuclear PTEN, the understanding of the mechanisms of its nuclear localization, function in the nucleus, and importance in biology and human disease has been lacking. Over the last decade, emerging concepts for the complex involvement of nuclear PTEN in a variety of processes, including genome maintenance and DNA repair, cell-cycle control, gene expression, and DNA replication, are illuminating what could prove to be the key path toward a full understanding of PTEN function in health and disease. Dysregulation of nuclear PTEN is now considered an important aspect of the etiology of many pathologic conditions, prompting reconsideration of the therapeutic approaches aimed at countering the consequences of PTEN deficiency. This new knowledge is fueling the development of innovative therapeutic modalities for a broad spectrum of human conditions, from cancer and metabolic diseases, to neurological disorders and autism.


Subject(s)
Cell Nucleus/metabolism , DNA Repair , PTEN Phosphohydrolase/metabolism , Animals , Apoptosis , Cell Cycle , DNA Replication , Humans , PTEN Phosphohydrolase/deficiency , Signal Transduction
6.
Breast ; 48: 17-23, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31472446

ABSTRACT

OBJECTIVES: Pre-clinical data suggest metformin might enhance the effect of chemotherapy in breast cancer (BC). We conducted a Phase II randomized trial of chemotherapy plus metformin versus placebo in metastatic breast cancer (MBC). MATERIAL AND METHODS: In this double blind phase II trial we randomly assigned non-diabetic MBC patients on 1st to 4th line chemotherapy to receive metformin 850 mg po bid or placebo bid. Primary outcome was progression-free survival (PFS); secondary outcomes included overall survival (OS), response rate (RR), toxicity and quality of life (QOL). With 40 subjects and a type-one error of 0.2 (one-sided), a PFS hazard ratio (HR) of 0.58 could be detected with 80% power. RESULTS: 40 patients were randomized (22 metformin, 18 placebo) with a mean age of 55 vs 57 years and ER/PR positive BC in 86.4% vs 83.3% off metformin vs placebo, respectively. Mean BMI was 27kg/m2 in both arms. The majority of patients were on 1st line chemotherapy. Grade 3-4 toxicity occurred in 31.8% (metformin) vs 58.8% (placebo). Best response: Partial response 18.2% metformin vs 25% placebo, stable disease 36.4% metformin vs 18.8% placebo, progressive disease 45.4% metformin vs 56.2% placebo. Mean PFS was 5.4 vs 6.3 months (metformin vs placebo), HR 1.2 (95% CI 0.63-2.31). Mean OS was 20.2 (metformin) vs 24.2 months (placebo), HR 1.68 (95% CI 0.79-3.55). CONCLUSION: In this population metformin showed no significant effect on RR, PFS or OS. These results do not support the use of metformin with chemotherapy in non-diabetic MBC patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Adult , Aged , Breast Neoplasms/pathology , Double-Blind Method , Female , Humans , Middle Aged , Progression-Free Survival , Quality of Life , Survival Rate
7.
Clin Cancer Res ; 25(17): 5398-5406, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31018921

ABSTRACT

PURPOSE: Exercise after breast cancer diagnosis is associated with lower cancer-specific mortality, but the biological mechanisms through which exercise impacts breast cancer are not fully understood. The Pre-Operative Health and Body (PreHAB) Study was a randomized window-of-opportunity trial designed to test the impact of exercise on Ki-67, gene expression, and other biomarkers in women with breast cancer. EXPERIMENTAL DESIGN: Inactive women with newly diagnosed breast cancer were randomized to an exercise intervention or mind-body control group, and participated in the study between enrollment and surgery (mean 29.3 days). Tumor and serum were collected at baseline and surgery. RESULTS: Forty-nine women were randomized (27 exercise, 22 control). At baseline, mean age was 52.6, body mass index was 30.2 kg/m2, and exercise was 49 minutes/week. Exercise participants significantly increased exercise versus controls (203 vs. 23 minutes/week, P < 0.0001). There were no differences in changes of expression of Ki-67, insulin receptor, and cleaved caspase-3 in exercise participants versus controls. KEGG pathway analysis demonstrated significant upregulation of 18 unique pathways between the baseline biopsy and surgical excision in exercise participants and none in control participants (q < 0.1). Top-ranked pathways included several implicated in immunity and inflammation. Exploratory analysis of tumor immune infiltrates demonstrated a trend toward a decrease in FOXP3+ cells in exercise versus control participants over the intervention period (P = 0.08). CONCLUSIONS: A window-of-opportunity exercise intervention did not impact proliferation but led to alterations in gene expression in breast tumors, suggesting that exercise may have a direct effect on breast cancer.See related commentary by Koelwyn and Jones, p. 5179.


Subject(s)
Breast Neoplasms , Cell Proliferation , Exercise , Exercise Therapy , Female , Humans , Preoperative Care
8.
JNCI Cancer Spectr ; 3(4): pkz049, 2019 Dec.
Article in English | MEDLINE | ID: mdl-32337478

ABSTRACT

Late disease recurrence (more than 5 years after initial diagnosis) represents a clinical challenge in the treatment and management of estrogen receptor-positive breast cancer (BC). An international workshop was convened in Toronto, Canada, in February 2018 to review the current understanding of late recurrence and to identify critical issues that require future study. The underlying biological causes of late recurrence are complex, with the processes governing cancer cell dormancy, including immunosurveillance, cell proliferation, angiogenesis, and cellular stemness, being integral to disease progression. These critical processes are described herein as well as their role in influencing risk of recurrence. Moreover, observational and interventional clinical trials are proposed, with a focus on methods to identify patients at risk of recurrence and possible strategies to combat this in patients with estrogen receptor-positive BC. Because the problem of late BC recurrence of great importance, recent advances in disease detection and patient monitoring should be incorporated into novel clinical trials to evaluate approaches to enhance patient management. Indeed, future research on these issues is planned and will offer new options for effective late recurrence treatment and prevention strategies.

9.
JNCI Cancer Spectr ; 3(4): pkz050, 2019 Dec.
Article in English | MEDLINE | ID: mdl-32337479

ABSTRACT

Disease recurrence (locoregional, distant) exerts a significant clinical impact on the survival of estrogen receptor-positive breast cancer patients. Many of these recurrences occur late, more than 5 years after original diagnosis, and represent a major obstacle to the effective treatment of this disease. Indeed, methods to identify patients at risk of late recurrence and therapeutic strategies designed to avert or treat these recurrences are lacking. Therefore, an international workshop was convened in Toronto, Canada, in February 2018 to review the current understanding of late recurrence and to identify critical issues that require future study. In this article, the major issues surrounding late recurrence are defined and current approaches that may be applicable to this challenge are discussed. Specifically, diagnostic tests with potential utility in late-recurrence prediction are described as well as a variety of patient-related factors that may influence recurrence risk. Clinical and therapeutic approaches are also reviewed, with a focus on patient surveillance and the implementation of extended endocrine therapy in the context of late-recurrence prevention. Understanding and treating late recurrence in estrogen receptor-positive breast cancer is a major unmet clinical need. A concerted effort of basic and clinical research is required to confront late recurrence and improve disease management and patient survival.

10.
JNCI Cancer Spectr ; 2(2): pky028, 2018 Apr.
Article in English | MEDLINE | ID: mdl-30035251

ABSTRACT

BACKGROUND: Circulating tumor cells (CTCs) are associated with worse prognosis in metastatic breast cancer (BC). We evaluated the association of metabolic, inflammatory, and tumor markers with CTCs in women with metastatic BC before commencing a new systemic therapy. METHODS: Ninety-six patients with newly diagnosed or progressing metastatic BC without current diabetes or use of anti-inflammatory agents were recruited from four Ontario hospitals. Women provided fasting blood for measurement of metabolic, inflammatory, and tumor markers and CTCs. CTCs were assayed within 72 hours of collection using CellSearch. Other blood was frozen at -80°C, and assays were performed in a single batch. Associations between CTC counts with study factors were evaluated using Spearman correlation, and the chi-square or Fisher exact test. All statistical tests were two-sided and P value ≤ .05 was considered statistically significant. RESULTS: The median age was 60.5 years; 90.6% were postmenopausal. The cohort included hormone receptor-positive (87.5%), HER2-positive (15.6%), and triple-negative (10.4%) BCs. Patients were starting firstline (35.5%), second-line (26.0%), or third-or-later-line therapy (38.5%). CTC counts (per 7.5 mL of blood) ranged from 0 to 1238 (median 2); an elevated CTC count, defined as five or more CTCs, was detected in 42 (43.8%) patients. Those with liver metastases (vs not) more frequently had an elevated CTC count (59.0% vs 33.3%, P = .02). CTCs were significantly associated with C-reactive protein (R = .22, P = .02), interleukin (IL)-6 (R = .25, P = .01), IL-8 (R = .38, P = .0001), plasminogen activator inhibitor 1 (R = .31, P = .001), carcinoembryonic antigen (R = .31, P = .002), and cancer antigen 15-3 (R = .40, P = .0001) and inversely associated with body mass index (R = -.23, P = .02) and leptin (R = -.26, P = .01). CONCLUSIONS: CTC counts were positively associated with tumor and inflammatory markers and inversely associated with some metabolic markers, potentially reflecting tumor burden and cachexia.

11.
J Clin Oncol ; 34(35): 4249-4255, 2016 12 10.
Article in English | MEDLINE | ID: mdl-27903146

ABSTRACT

Over the past 40 years, the prevalence of obesity has increased epidemically worldwide, which raises significant concerns regarding public health and the associated economic burden. Obesity is a major risk factor for several conditions including cardiovascular disease and type 2 diabetes, and recent evidence suggests that obesity negatively affects cancer risk and outcome. The relationship between obesity and cancer is complex and involves multiple factors both at the systemic and cellular level. Indeed, disruptions in insulin metabolism, adipokines, inflammation, and sex hormones all contribute to the adverse effects of obesity in cancer development and progression. The focus of this review will be the impact of these systemic obesity-related factors on cancer biology, incidence, and outcome. Potential therapeutic interventions and current clinical trials targeting obesity and its associated factors will also be discussed.


Subject(s)
Energy Metabolism , Neoplasms/complications , Neoplasms/metabolism , Obesity/complications , Obesity/metabolism , Adipokines/metabolism , Adipose Tissue/metabolism , Humans , Inflammation/metabolism , Insulin Resistance , Risk Factors
13.
Breast Cancer Res ; 17: 32, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25849721

ABSTRACT

INTRODUCTION: The antidiabetic drug metformin exhibits potential anticancer properties that are believed to involve both direct (insulin-independent) and indirect (insulin-dependent) actions. Direct effects are linked to activation of AMP-activated protein kinase (AMPK) and an inhibition of mammalian target of rapamycin mTOR signaling, and indirect effects are mediated by reductions in circulating insulin, leading to reduced insulin receptor (IR)-mediated signaling. However, the in vivo impact of metformin on cancer cell signaling and the factors governing sensitivity in patients remain unknown. METHODS: We conducted a neoadjuvant, single-arm, "window of opportunity" trial to examine the clinical and biological effects of metformin on patients with breast cancer. Women with untreated breast cancer who did not have diabetes were given 500 mg of metformin three times daily for ≥2 weeks after diagnostic biopsy until surgery. Fasting blood and tumor samples were collected at diagnosis and surgery. Blood glucose and insulin were assayed to assess the physiologic effects of metformin, and immunohistochemical analysis of tumors was used to characterize cellular markers before and after treatment. RESULTS: Levels of IR expression decreased significantly in tumors (P = 0.04), as did the phosphorylation status of protein kinase B (PKB)/Akt (S473), extracellular signal-regulated kinase 1/2 (ERK1/2, T202/Y204), AMPK (T172) and acetyl coenzyme A carboxylase (S79) (P = 0.0001, P < 0.0001, P < 0.005 and P = 0.02, respectively). All tumors expressed organic cation transporter 1, with 90% (35 of 39) exhibiting an Allred score of 5 or higher. CONCLUSIONS: Reduced PKB/Akt and ERK1/2 phosphorylation, coupled with decreased insulin and IR levels, suggest insulin-dependent effects are important in the clinical setting. These results are consistent with beneficial anticancer effects of metformin and highlight key factors involved in sensitivity, which could be used to identify patients with breast cancer who may be responsive to metformin-based therapies. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT00897884. Registered 8 May 2009.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Metformin/pharmacology , Receptor, Insulin/metabolism , Signal Transduction/drug effects , Adult , Aged , Antineoplastic Agents/therapeutic use , Biomarkers/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Female , Humans , Metformin/therapeutic use , Middle Aged , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoadjuvant Therapy , Neoplasm Grading , Neoplasm Staging , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
14.
Breast Cancer Res Treat ; 150(1): 149-55, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25682077

ABSTRACT

Metformin has therapeutic potential against breast cancer, but the mechanisms of action in vivo remain uncertain. This study examined biomarker effects of metformin in primary breast cancer in a preoperative window of opportunity trial. Non-diabetic women with operable invasive breast cancer were randomized to receive open label pre-operative metformin (500 mg daily for 1 week then 1 g twice daily for a further week) or as controls, not receiving metformin. Patients in both arms had a core biopsy pre-randomisation and again at the time of surgery. Immunohistochemistry for phospho-AMPK (pAMPK), phospho-Akt (pAkt), insulin receptor, cleaved caspase-3, and Ki67 was performed on formalin-fixed paraffin-embedded cores, scored blinded to treatment and analysed by paired t test. In metformin-treated patients, significant up-regulation of pAMPK (paired t test, p = 0.04) and down-regulation of pAkt (paired t test, p = 0.043) were demonstrated compared to the control group. Insulin receptor and serum insulin remained similar following metformin treatment compared with a rise in insulin receptor and insulin in controls. Significant falls in Ki67 and cleaved caspase-3 (paired t test, p = 0.044) were seen in the metformin-treated patients but not in the control group. Changes were independent of body mass index. These biomarker data suggest mechanisms for metformin action in vivo in breast cancer patients via up-regulation of tumor pAMPK, down-regulation of pAkt, and suppression of insulin responses reflecting cytostatic rather than cytotoxic mechanisms.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Metformin/therapeutic use , Antineoplastic Agents/administration & dosage , Biomarkers/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 3/metabolism , Female , Humans , Ki-67 Antigen/metabolism , Metformin/administration & dosage , Neoadjuvant Therapy , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Signal Transduction , Treatment Outcome
15.
Breast Cancer Res Treat ; 135(3): 821-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22933030

ABSTRACT

Metformin may exert anti-cancer effects through indirect (insulin-mediated) or direct (insulin-independent) mechanisms. We report results of a neoadjuvant "window of opportunity" study of metformin in women with operable breast cancer. Newly diagnosed, untreated, non-diabetic breast cancer patients received metformin 500 mg tid after diagnostic core biopsy until definitive surgery. Clinical (weight, symptoms, and quality of life) and blood [fasting serum insulin, glucose, homeostasis model assessment (HOMA), C-reactive protein (CRP), and leptin] attributes were compared pre- and post-metformin as were terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) and Ki67 scores (our primary endpoint) in tumor tissue. Thirty-nine patients completed the study. Mean age was 51 years, and metformin was administered for a median of 18 days (range 13-40) up to the evening prior to surgery. 51 % had T1 cancers, 38 % had positive nodes, 85 % had ER and/or PgR positive tumors, and 13 % had HER2 overexpressing or amplified tumors. Mild, self-limiting nausea, diarrhea, anorexia, and abdominal bloating were present in 50, 50, 41, and 32 % of patients, respectively, but no significant decreases were seen on the EORTC30-QLQ function scales. Body mass index (BMI) (-0.5 kg/m(2), p < 0.0001), weight (-1.2 kg, p < 0.0001), and HOMA (-0.21, p = 0.047) decreased significantly while non-significant decreases were seen in insulin (-4.7 pmol/L, p = 0.07), leptin (-1.3 ng/mL, p = 0.15) and CRP (-0.2 mg/L, p = 0.35). Ki67 staining in invasive tumor tissue decreased (from 36.5 to 33.5 %, p = 0.016) and TUNEL staining increased (from 0.56 to 1.05, p = 0.004). Short-term preoperative metformin was well tolerated and resulted in clinical and cellular changes consistent with beneficial anti-cancer effects; evaluation of the clinical relevance of these findings in adequately powered clinical trials using clinical endpoints such as survival is needed.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Apoptosis/drug effects , Body Mass Index , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Ki-67 Antigen/analysis , Metformin/administration & dosage , Metformin/adverse effects , Middle Aged , Neoadjuvant Therapy , Preoperative Care , Quality of Life
16.
Nat Immunol ; 13(6): 543-550, 2012 Apr 29.
Article in English | MEDLINE | ID: mdl-22544393

ABSTRACT

Type I interferon is an integral component of the antiviral response, and its production is tightly controlled at the levels of transcription and translation. The eukaryotic translation-initiation factor eIF4E is a rate-limiting factor whose activity is regulated by phosphorylation of Ser209. Here we found that mice and fibroblasts in which eIF4E cannot be phosphorylated were less susceptible to virus infection. More production of type I interferon, resulting from less translation of Nfkbia mRNA (which encodes the inhibitor IκBα), largely explained this phenotype. The lower abundance of IκBα resulted in enhanced activity of the transcription factor NF-κB, which promoted the production of interferon-ß (IFN-ß). Thus, regulated phosphorylation of eIF4E has a key role in antiviral host defense by selectively controlling the translation of an mRNA that encodes a critical suppressor of the innate antiviral response.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Interferon Type I/biosynthesis , NF-kappa B/metabolism , Vesicular Stomatitis/immunology , Vesicular stomatitis Indiana virus/physiology , Animals , Electrophoretic Mobility Shift Assay , Eukaryotic Initiation Factor-4E/immunology , Female , I-kappa B Proteins/biosynthesis , I-kappa B Proteins/genetics , I-kappa B Proteins/immunology , Immunity, Innate/immunology , Immunoblotting , Interferon Type I/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-KappaB Inhibitor alpha , NF-kappa B/immunology , Phosphorylation , Protein Biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms , Vesicular Stomatitis/genetics , Vesicular Stomatitis/metabolism , Vesicular Stomatitis/virology , Vesicular stomatitis Indiana virus/immunology , Virus Replication
17.
J Mol Endocrinol ; 48(3): R31-43, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22355097

ABSTRACT

The anti-diabetic drug metformin is rapidly emerging as a potential anti-cancer agent. Metformin, effective in treating type 2 diabetes and the insulin resistance syndromes, improves insulin resistance by reducing hepatic gluconeogenesis and by enhancing glucose uptake by skeletal muscle. Epidemiological studies have consistently associated metformin use with decreased cancer incidence and cancer-related mortality. Furthermore, numerous preclinical and clinical studies have demonstrated anti-cancer effects of metformin, leading to an explosion of interest in evaluating this agent in human cancer. The effects of metformin on circulating insulin levels indicate a potential efficacy towards cancers associated with hyperinsulinaemia; however, metformin may also directly inhibit tumour growth. In this review, we describe the mechanism of action of metformin and summarise the epidemiological, clinical and preclinical evidence supporting a role for metformin in the treatment of cancer. In addition, the challenges associated with translating preclinical results into therapeutic benefit in the clinical setting will be discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Metformin/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Metformin/metabolism , Neoplasms/metabolism , Translational Research, Biomedical
18.
BMC Med ; 9: 33, 2011 Apr 06.
Article in English | MEDLINE | ID: mdl-21470407

ABSTRACT

Biguanides have been developed for the treatment of hyperglycemia and type 2 diabetes. Recently, metformin, the most widely prescribed biguanide, has emerged as a potential anticancer agent. Epidemiological, preclinical and clinical evidence supports the use of metformin as a cancer therapeutic. The ability of metformin to lower circulating insulin may be particularly important for the treatment of cancers known to be associated with hyperinsulinemia, such as those of the breast and colon. Moreover, metformin may exhibit direct inhibitory effects on cancer cells by inhibiting mammalian target of rapamycin (mTOR) signaling and protein synthesis. The evidence supporting a role for metformin in cancer therapy and its potential molecular mechanisms of action are discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Metformin/therapeutic use , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Drug Therapy, Combination/methods , Humans , Metformin/pharmacology
19.
Science ; 328(5982): 1172-6, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20508131

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) integrates mitogen and nutrient signals to control cell proliferation and cell size. Hence, mTORC1 is implicated in a large number of human diseases--including diabetes, obesity, heart disease, and cancer--that are characterized by aberrant cell growth and proliferation. Although eukaryotic translation initiation factor 4E-binding proteins (4E-BPs) are critical mediators of mTORC1 function, their precise contribution to mTORC1 signaling and the mechanisms by which they mediate mTORC1 function have remained unclear. We inhibited the mTORC1 pathway in cells lacking 4E-BPs and analyzed the effects on cell size, cell proliferation, and cell cycle progression. Although the 4E-BPs had no effect on cell size, they inhibited cell proliferation by selectively inhibiting the translation of messenger RNAs that encode proliferation-promoting proteins and proteins involved in cell cycle progression. Thus, control of cell size and cell cycle progression appear to be independent in mammalian cells, whereas in lower eukaryotes, 4E-BPs influence both cell growth and proliferation.


Subject(s)
Carrier Proteins/metabolism , Cell Enlargement , Cell Proliferation , Eukaryotic Initiation Factors/metabolism , Phosphoproteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Cell Cycle , Cell Cycle Proteins , Cell Line , Cell Size , Cell Survival , Eukaryotic Initiation Factors/genetics , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes , Phosphoproteins/genetics , Phosphorylation , Protein Biosynthesis , Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
20.
Bioorg Med Chem ; 18(22): 7746-52, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20427189

ABSTRACT

Spatially localized translation plays a vital role in the normal functioning of neuronal systems and is widely believed to be involved in both learning and memory formation. It is of central interest to understand both the phenomenon and molecular mechanisms of local translation using new tools and approaches. Caged compounds can, in principle, be used as tools to investigate local translation since optical activation of bioactive molecules can achieve both spatial and temporal resolution on the micron scale and on the order of seconds or less, respectively. Successful caging of bioactive molecules requires the identification of key functional groups in appropriate molecules and the introduction of a suitable caging moiety. Here we present the design, synthesis and testing of a collection of three caged compounds: anisomycin caged with a diethylaminocoumarin moiety and dimethoxynitrobenzyl caged versions of 4E-BP and rapamycin. Whereas caged anisomycin can be used to control general translation, caged 4E-BP serves as a probe of cap-dependent translation initiation and caged rapamycin serves a probe of the role of mTORC1 in translation initiation. In vitro translation assays demonstrate that these caging strategies, in combination with the aforementioned compounds, are effective for optical control making it likely that such strategies can successfully employed in the study of local translation in living systems.


Subject(s)
Aminocoumarins/chemistry , Nervous System/metabolism , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/chemistry , Animals , Anisomycin/pharmacology , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Protein Synthesis Inhibitors/pharmacology , Proteins/antagonists & inhibitors , Proteins/metabolism , RNA/metabolism , Rabbits , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...